parkin structure and function

13
STATE-OF-THE-ART REVIEW Parkin structure and function Marjan Seirafi, Guennadi Kozlov and Kalle Gehring Department of Biochemistry and the Groupe de Recherche ax e sur la Structure des Prot eines, McGill University, Montr eal, Qu ebec, Canada Keywords mitophagy; parkin; PINK1; ubiquitin; ubiquitination Correspondence K. Gehring, Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Montr eal, Qu ebec, Canada H3G 0B1 Fax: 514 398-2983 Tel: 514 398-7287 E-mail: [email protected] (Received 4 October 2014, revised 5 January 2015, accepted 24 February 2015) doi:10.1111/febs.13249 Mutations in the parkin or PINK1 genes are the leading cause of the auto- somal recessive form of Parkinson’s disease. The gene products, the E3 ubiquitin ligase parkin and the serine/threonine kinase PINK1, are neuro- protective proteins, which act together in a mitochondrial quality control pathway. Here, we review the structure of parkin and mechanisms of its autoinhibition and function as a ubiquitin ligase. We present a model for the recruitment and activation of parkin as a key regulatory step in the clearance of depolarized or damaged mitochondria by autophagy (mito- phagy). We conclude with a brief overview of other functions of parkin and considerations for drug discovery in the mitochondrial quality control pathway. Introduction Parkinson’s disease (PD) is a degenerative movement disorder caused by the progressive death of dopamine- producing neurons in the substantia nigra pars com- pacta of the mid-brain. Studies linking PD to defects in the electron transport chain suggest that damaged mitochondria may play a central role in PD pathology [1]. While most PD cases occur sporadically, research on inherited forms of PD in the past decade have shed new light on the disease and pathogenesis. Studies of two recessive PD genes, PINK1 (PTEN-induced puta- tive kinase protein 1 or PARK6) and parkin (PARK2), have provided direct evidence for the con- tribution of damaged mitochondria in PD pathology [2]. Parkin is a cytosolic E3 ubiquitin ligase and PINK1 is the only known protein kinase with a mitochondrial targeting domain. These two proteins are involved in a common pathway regulating mitochondrial quality control and promoting the selec- tive autophagy of depolarized mitochondria (mito- phagy) [3]. Under basal conditions, parkin E3 ligase activity is repressed in the cytoplasm and PINK1 is imported into mitochondria via TOM40 and TOM20 core containing complexes [4] and cleaved sequentially by mitochondrial proteases such as MPP and PARL [58]. Cleaved PINK1 then degrades rapidly in the cytosol via the N-end rule pathway so that levels of PINK1 are low in healthy cells [9]. However, when the organelle loses its inner membrane electrochemical gra- dient, import of PINK1 to the mitochondria is inhib- ited and the protein is stabilized on the mitochondrial outer membrane with its kinase domain on the cyto- solic face [10,11]. The accumulation of PINK1 kinase activity on the mitochondria triggers parkin recruit- ment and activation. Activated parkin produces Abbreviations CCCP, carbonyl cyanide m-chlorophenyl hydrazone; HECT, homologous to the E6-AP carboxyl terminus; IBR, in-between-RING; PD, Parkinson’s disease; RBR, RING-between-RING; REP, repressor element of parkin; RING, really interesting new gene; ROS, reactive oxygen species; Ubl, ubiquitin-like domain; UIM, ubiquitin interacting motif. 2076 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS. This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.

Upload: vuongxuyen

Post on 04-Jan-2017

233 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Parkin structure and function

STATE-OF-THE-ART REVIEW

Parkin structure and functionMarjan Seirafi, Guennadi Kozlov and Kalle Gehring

Department of Biochemistry and the Groupe de Recherche ax�e sur la Structure des Prot�eines, McGill University, Montr�eal, Qu�ebec, Canada

Keywords

mitophagy; parkin; PINK1; ubiquitin;

ubiquitination

Correspondence

K. Gehring, Department of Biochemistry,

McGill University, 3649 Promenade Sir

William Osler, Montr�eal, Qu�ebec, Canada

H3G 0B1

Fax: 514 398-2983

Tel: 514 398-7287

E-mail: [email protected]

(Received 4 October 2014, revised 5

January 2015, accepted 24 February 2015)

doi:10.1111/febs.13249

Mutations in the parkin or PINK1 genes are the leading cause of the auto-

somal recessive form of Parkinson’s disease. The gene products, the E3

ubiquitin ligase parkin and the serine/threonine kinase PINK1, are neuro-

protective proteins, which act together in a mitochondrial quality control

pathway. Here, we review the structure of parkin and mechanisms of its

autoinhibition and function as a ubiquitin ligase. We present a model for

the recruitment and activation of parkin as a key regulatory step in the

clearance of depolarized or damaged mitochondria by autophagy (mito-

phagy). We conclude with a brief overview of other functions of parkin

and considerations for drug discovery in the mitochondrial quality control

pathway.

Introduction

Parkinson’s disease (PD) is a degenerative movement

disorder caused by the progressive death of dopamine-

producing neurons in the substantia nigra pars com-

pacta of the mid-brain. Studies linking PD to defects

in the electron transport chain suggest that damaged

mitochondria may play a central role in PD pathology

[1]. While most PD cases occur sporadically, research

on inherited forms of PD in the past decade have shed

new light on the disease and pathogenesis. Studies of

two recessive PD genes, PINK1 (PTEN-induced puta-

tive kinase protein 1 or PARK6) and parkin

(PARK2), have provided direct evidence for the con-

tribution of damaged mitochondria in PD pathology

[2]. Parkin is a cytosolic E3 ubiquitin ligase and

PINK1 is the only known protein kinase with a

mitochondrial targeting domain. These two proteins

are involved in a common pathway regulating

mitochondrial quality control and promoting the selec-

tive autophagy of depolarized mitochondria (mito-

phagy) [3]. Under basal conditions, parkin E3 ligase

activity is repressed in the cytoplasm and PINK1 is

imported into mitochondria via TOM40 and TOM20

core containing complexes [4] and cleaved sequentially

by mitochondrial proteases such as MPP and PARL

[5–8]. Cleaved PINK1 then degrades rapidly in the

cytosol via the N-end rule pathway so that levels of

PINK1 are low in healthy cells [9]. However, when the

organelle loses its inner membrane electrochemical gra-

dient, import of PINK1 to the mitochondria is inhib-

ited and the protein is stabilized on the mitochondrial

outer membrane with its kinase domain on the cyto-

solic face [10,11]. The accumulation of PINK1 kinase

activity on the mitochondria triggers parkin recruit-

ment and activation. Activated parkin produces

Abbreviations

CCCP, carbonyl cyanide m-chlorophenyl hydrazone; HECT, homologous to the E6-AP carboxyl terminus; IBR, in-between-RING; PD,

Parkinson’s disease; RBR, RING-between-RING; REP, repressor element of parkin; RING, really interesting new gene; ROS, reactive oxygen

species; Ubl, ubiquitin-like domain; UIM, ubiquitin interacting motif.

2076 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and

distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.

Page 2: Parkin structure and function

ubiquitin chains on various outer mitochondrial

membrane proteins leading to autophagic elimination

of the damaged organelle [12–15]. Pathogenic muta-

tions in either of these genes lead to loss of this quality

control pathway and accumulation of impaired

mitochondria, which are thought to be a source of

toxic reactive oxygen species (ROS) and contribute to

neuronal cell death and PD.

Parkin is an RBR E3 ubiquitin ligase

Ubiquitination is a post-translational modification that

typically marks proteins for degradation through the

covalent attachment of ubiquitin and ubiquitin chains

to lysine residues or the N-terminal amino group of a

substrate protein [16]. In addition to degradation via

the proteasome, ubiquitination can act as a signal for

autophagy [17] – degradation via lysosomes – as well

as alter substrate protein activity or location [18].

Ubiquitination is carried out through the sequential

action of three enzymes: E1 ubiquitin-activating

enzymes, E2 ubiquitin-conjugating enzymes and E3

ubiquitin ligases (Fig. 1A). In the pathway, E1 first

uses ATP to activate ubiquitin for conjugation by

forming a thioester between its catalytic cysteine and

the C-terminal carboxyl group of ubiquitin. The

ubiquitin is then passed to a second cysteine of an E2

ubiquitin conjugating enzyme. In the final step, the

ubiquitin-charged E2 enzyme interacts with a specific

E3 ubiquitin ligase and transfers the ubiquitin to the

amino group of a substrate protein. Ubiquitin contains

seven lysine residues as well as an N-terminal amino

group that can be used to build chains of polyubiqu-

itin. The most common chain types are K48 and K63

chains in which multiple ubiquitin molecules are linked

in a linear arrangement with the C terminus of one

molecule attached to lysine 48 (or 63) of the next. In

the ubiquitination pathway, the E3 ubiquitin ligases

typically provide the majority of the specificity and

regulation in recognition of substrates and control of

activity [19]. Based on considerations of structure and

chemistry, three classes of E3 ligases are distinguished:

RING-type (including U-box ligases), HECT-type and

RING-HECT hybrids [20] (Fig. 1B). RING-type E3s

are characterized by the presence of a canonical

C3HC4-type RING (really interesting new gene)

domain that binds the E2 enzyme but does not partici-

pate directly in catalysis. This class of E3s functions as

inert scaffolding ligases that facilitate the direct trans-

fer of ubiquitin from the E2 onto the substrate.

HECT-type E3s contain a HECT (homologous to the

E6-AP carboxyl terminus) domain with an active site

cysteine, which accepts ubiquitin from an E2 enzyme

in the form of a thioester intermediate and then trans-

fers it to substrates. The third class consists of the

RBR (RING-between-RING) family of E3 ubiquitin

ligases that combine the chemistry of HECT-type lig-

ases with structural similarity to RING-type ligases

[21,22]. RBRs contain a canonical C3HC4-type RING

(named RING1), followed by two conserved Cys/His-

rich Zn-binding domains, in-between-RING (IBR) and

RING2 domains, that contain an active site cysteine

residue. This cysteine accepts ubiquitin from the E2

enzyme and transfers it onto substrates; hence, ligases

in this class are sometimes referred to as RING-HECT

hybrids [22].

Parkin, a member of RBR E3 ubiquitin ligases,

ubiquitinates a wide variety of cytosolic and outer

mitochondrial membrane proteins upon mitochondrial

depolarization [23,24]. It forms multiple types of

ubiquitin chains, most frequently K63, K48, K11 and

K6 linkages [25]. Parkin also shows relatively lax sub-

strate specificity [23,24]. Accumulation of polyubiqu-

itin chains on mitochondria signals recruitment of the

autophagosome and proteasome machinery to initiate

mitophagy [12–14,23]. Parkin itself becomes ubiquiti-

nated by the attachment of K6 ubiquitin chains, which

may play a role in its own degradation [26]. The activ-

ity of parkin is tightly regulated and normally

repressed [27–30]. In cells, parkin activity can be acti-

vated under a variety of conditions such as depolariza-

tion of mitochondria or epidermal growth factor

signaling [31]. A large number of treatments that dis-

rupt its structure are also known to de-repress its

ligase activity in vitro. These include heat treatment,

N-terminal deletions and some point mutations [28–30,32].

Lessons from the structure

Recent structures of parkin have revealed much about

its regulation and function (Fig. 2A). Parkin consists

of a ubiquitin-like domain (Ubl) at its N terminus and

four zinc-coordinating RING-like domains: RING0,

RING1, IBR and RING2. More than 120 pathogenic

PD mutations are spread throughout parkin domains,

attesting to critical functions for each of the individual

domains [33]. The Ubl domain is involved in substrate

recognition, binding SH3 and ubiquitin interacting

motif (UIM) domains, proteasome association, and

regulation of cellular parkin levels and activity

[27,31,34–37].Last year, several groups reported high-resolution

crystal structures of a parkin fragment consisting of

the RING0, RING1, IBR and RING2 domains (PDB:

4K7D, 4I1H, 4I1F, 4BM9) along with a low-resolution

2077FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

M. Seirafi et al. Parkin structure and function

Page 3: Parkin structure and function

structure of the full-length protein (PDB: 4K95) [28–30,38]. In the structures, parkin adopts a compact

arrangement, stabilized by multiple hydrophobic inter-

actions, similar to a coiled snake [39] (Fig. 2B). The

N-terminal Ubl domain uses a hydrophobic surface

centered around Ile44 to bind to the RING1 domain,

while the C-terminal catalytic domain is tightly associ-

ated with the RING0 domain. Domains RING0

through RING2 (collectively referred to as R0–RBR)

each coordinate two zinc ions through histidine and

cysteine residues, confirming the stoichiometry of eight

zinc ions per parkin [40]. The RING0 domain binds

zinc ions in a hairpin arrangement unique to parkin

while the RING2 and IBR domains show a sequential

arrangement of zinc-coordinating residues. RING0

and RING2 were originally identified as RING

domains based on the primary amino acid sequence

but their structural topology differs from a classical

RING fold. The RING1 domain is the only RING

domain with the cross-brace zinc coordination topol-

ogy observed in other RING-type E3 ligases. The simi-

larity of RING1 to other RING E3 ligases suggested

that it is the E2 binding site on parkin (Fig. 2C).

NMR titrations, mutagenesis and molecular modeling

ATP

E1

S

UbCO

UbCO

O-

Substrate

E2

S

UbCO

E3

Substrate

E2

S

UbCO

E2

SHE1

SH

E3

Substrate

NH

UbCO Ub Ub Ub

Substrate

NH

UbCO

UbUb

Ub

UbUb

UbUb

Substrate

NH

UbCO

Mono-ubiquitinationEndocytosis

K48 polyubiquitinationProteosomal & lysosomal degradation

Substrate

NH

CO

Linear chain polyubiquitinationNF-kB signalling

K63 polyubiquitinationSignalling, DNA repair

Lysosomal degradation

NH2

Substrate

E2

S

UbCO

RING

Substrate

E2

S

UbCO

NH2

RING

HECT E3E2

S

UbCO SH

E2

S

UbCO

SH

RINGE3

N-lope

C-lope

N-lope

C-lopeSubstrateSubstrate

NH2

RING1 IBR RING2RING/HECT hybrid

E3Substrate

RING1 IBR RING2 Substrate

NH2

A

B

Fig. 1. Ubiquitination pathway. (A) Cascade of ubiquitination enzymes. The ubiquitin-activating enzyme E1 uses ATP to conjugate the C-

terminal carboxylic acid group of ubiquitin to an active site cysteine. This is then transferred to a cysteine on one of a number of E2

enzymes that work in concert with E3 enzymes to ubiquitinate substrate proteins on amino groups of lysine residues or protein N termini.

The formations of mono-ubiquitin or polyubiquitin chains on substrates are signals for different downstream pathways. (B) Classes of E3

ubiquitin ligases. E3 ligases can be distinguished based on the presence of a RING domain, a catalytic cysteine or both. RING and U-box

domain ligases act as scaffolds to bring the substrate and ubiquitin-conjugated E2 together. HECT ligases have a catalytic cysteine in their

C-lobe that is transiently conjugated to ubiquitin as an intermediate in a two-step process of substrate ubiquitination. Parkin is a RING/HECT

hybrid ligase that contains a RING domain that binds the E2 enzyme and a catalytic cysteine that transfers ubiquitin to the substrate.

2078 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

Parkin structure and function M. Seirafi et al.

Page 4: Parkin structure and function

confirmed this and identified an a-helix (residues 263–271) and two loops (residues 239–244 and 290–292) inRING1 as the E2 binding site [28–30]. The RING2

domain is the catalytic module harboring the catalytic

cysteine (Cys431). While RING0 is unique to parkin,

the IBR domain is conserved among the RBR E3 fam-

ily; the precise function of the domains is currently

unknown. Parkin contains two interdomain linkers

that are flexible and not observed in the crystal struc-

tures. The first consists of 70 poorly conserved residues

of unknown function that follow the Ubl domain. The

second, within the R0–RBR fragment, occurs between

the IBR and RING2 domains and is composed of dis-

ordered segments and a conserved a-helix (residues

391–403) that binds to the RING1 domain. This helix

has been termed the repressor element of parkin

(REP) due to its role in the regulation of parkin activ-

ity.

Parkin is a difficult protein to work with unless cer-

tain precautions are taken [41]. The ligase activity is

normally repressed and surprisingly some mutations or

heat treatment increase activity [27–30]. The bound

zinc ions are required for structural stability and prop-

erly regulated enzymatic activity [40,42]. While supple-

mental zinc is not required during purification, metal-

chelating agents such as ethylenediaminetetraacetic

acid (EDTA) need to be avoided since they denature

the protein. Unfortunately, some published co-immu-

noprecipitation experiments have used EDTA and

need to be interpreted with caution. With a total of 35

cysteine residues, parkin requires a high concentration

of a reducing agent such as dithiothreitol to maintain

the cysteine thiols reduced and available for coordinat-

ing zinc ions.

The parkin structure provides rationales for many

of the mutations associated with PD (Fig. 2A). Certain

mutations compromise the structural integrity of the

protein. Others interfere with binding of substrates or

directly affect the enzyme catalysis. Among the best

understood, mutations C212Y, C289G and C441R

affect zinc coordination, while R42P, K211N and

T351P disrupt protein folding or stability. C431F and

G430D are in the catalytic site, and T240R prevents

E2 binding. The effects of other mutations such as

R33Q, D280N, G328E or T415N are less clear. These

may disrupt the interdomain interactions or affect

IBR

E2 enzyme

RING0

RING2

RING1

Cys431

Cysteine

R33Q R42P V65E K161N

M192L C212Y R234QT240M

R275WD280N

C289GG328E

R334CT351P T415N

G430DC431F

E444QW453s top

Ubl RING0 RING1 IBR RING2

1 76 141 225 327 378 410 465

REP

A

A398T

IBR

RING0

RING2

RING1

REPUbl

Phospho-siteSer65

Catalytic siteCys431

B C

K211N

Fig. 2. The structure of parkin reveals the mechanism of its autoinhibition. (A) Domain architecture of the five parkin domains and

identification of selected PD mutations. (B) Cartoon representation of parkin (PDB: 4K95) [29]. Parkin activity depends on two functional

sites: a binding site for ubiquitin-conjugated E2 enzyme on the RING1 of parkin and a catalytic site with a cysteine that forms a transient

covalent linkage with ubiquitin on the RING2 domain. Both sites are occluded in the autoinhibited structure. The Ubl domain and REP linker

between the IBR and RING2 domains prevent the E2 from binding to RING1. The RING0 domain partially covers the catalytic cysteine on

the RING2 domain. Mutations at interdomain interfaces as well as phosphorylation of Ser65 in the Ubl domain increase parkin ubiquitin

ligase activity. The eight structural zinc ions in parkin are shown as gray spheres. Dashed lines indicate portions of the IBR and RING2 linker

that were not observed in the crystal structure. (C) Model of an E2 enzyme bound to parkin with Ubl and REP linker removed. An additional

structural rearrangement must occur to allow ubiquitin on the E2 enzyme to transfer to the catalytic cysteine of parkin. The E2 and parkin

catalytic cysteines are ~ 50 �A apart in the model.

2079FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

M. Seirafi et al. Parkin structure and function

Page 5: Parkin structure and function

binding of other proteins by parkin. A comprehensive

list of known disease-causing parkin mutations

together with the predicted structure rationale and bio-

chemical effects is provided as supplementary material

by Wauer and Komander [30].

Autoinhibition

The activity of parkin is tightly controlled by multiple

mechanisms of autoinhibition. The first is access to the

catalytic RING2 domain, which is blocked by RING0.

Other RBR E3 ligases show similar mechanisms of au-

toinhibition [43–45]. The Ariadne domain of the RBR

E3 HHARI and the UBA domain of the RBR E3

HOIP both participate in inhibiting the catalytic

domain. Deletion of the N terminus of parkin through

to the RING0 domain leads to very high ubiquitin

ligase activity in vitro [28–30].A second mechanism is the control of binding of the

upstream E2 enzyme to parkin. Modeling and muta-

genesis have confirmed that the E2 binding site is on

the RING1 domain but the site is occluded by the Ubl

domain and REP linker (Fig. 2C). Deletion of the Ubl

domain and mutagenesis of the REP linker both

increase the affinity of E2 binding and parkin activity

[27,29]. Finally, the large distance between the E2

binding site and catalytic site on RING2 prevents

transfer of ubiquitin from the ubiquitin–E2 conjugate

to the parkin catalytic cysteine (Fig. 2C).

Catalytic mechanism

The detailed structure of parkin RING2 domain

reveals conserved chemistry across the RBR family of

E3 ligases for ubiquitin transfer to the target protein

(Fig. 3). Catalytically, ubiquitination proceeds

through two steps: (a) formation of a ubiquitin–cyste-ine thioester intermediate where the C terminus of

ubiquitin is covalently linked to the parkin catalytic

cysteine, and (b) acyl transfer where the ubiquitin is

transferred from parkin to an amino group of the

substrate.

A C

B

Fig. 3. Mechanism of parkin ubiquitin ligase activity. (A) Sequence alignment of RBR E3 ubiquitin ligases shows conservation in the catalytic

RING2 domains. The catalytic cysteine (highlighted in yellow) is invariant across RBR proteins, whereas histidine (green) and glutamate

residues (red) that play secondary roles in catalysis are less conserved. Gray indicates zinc-coordinating residues. (B) Structural comparison

of catalytic domains of parkin, HHARI and HOIP (PDB: 4K7D [29]; 4KBL [43]; 4LJO [44]). The alignment of the catalytic cysteine, histidine

and glutamate/glutamine is suggestive of a catalytic triad where the histidine acts as a general base to promote transesterification of

ubiquitin onto the substrate. (C) Model of the positions of the donor and acceptor ubiquitin molecules in the catalytic site of parkin based on

the crystal structure of HOIP [44]. The C terminus of the donor ubiquitin lies in a groove of the RING2 domain and terminates next to the

catalytic cysteine, mimicking the thioester intermediate. The amino group of the acceptor ubiquitin approaches Cys431 from the opposite

side but would be sterically blocked by the parkin RING0 domain in the absence of a conformational change.

2080 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

Parkin structure and function M. Seirafi et al.

Page 6: Parkin structure and function

The catalytic cysteine residue (in the RING2

domain) is conserved in all the members of the RBR

family as are the residues involved in zinc coordina-

tion. Following the cysteine, there is partial conserva-

tion of a histidine residue and an acidic residue. In

parkin, these residues adopt a linear arrangement that

has been proposed to act as a catalytic triad where the

histidine acts as a general base to promote catalysis

[28–30]. The structures of two other RBR ligases are

known: HHARI, a member of the Ariadne family of

E3 ligases [43], and HOIP, a component of the linear

ubiquitination assembly complex (LUBAC) [44]. The

RING2 domains are highly conserved and share the

linear arrangement of the three residues (Fig. 3B).

Nonetheless, the results of mutagenesis are ambiguous

about the importance of the catalytic triad. The effect

of the loss of the histidine is substrate dependent and

can be suppressed in vitro by raising the pH [28,44]. In

cultured cells, mutation of the histidine only moder-

ately slows parkin-mediated mitophagy. And, while

the mutation E444Q reduces autoubiquitination [29]

and may be implicated in PD [46], it has no effect on

parkin activity in cells [28].

Additional insight into the catalytic mechanism

comes from the crystal structure of HOIP, which con-

tains two ubiquitin molecules in contact with catalytic

RING2 domain (Fig. 3C). The C terminus of one

ubiquitin molecule is positioned to mimic the thioester

linkage with the catalytic cysteine while the amino

group of the second ubiquitin molecule approaches the

cysteine from the other side and occupies the position

of the acceptor molecule. Transposition of the two

ubiquitin molecules onto the parkin crystal structure

generates a hypothetical model of the active site with a

thioester intermediate prior to acyl transfer. While the

donor ubiquitin can be accommodated to fit in a

groove on the surface of the RING2 domain, the posi-

tion of the acceptor ubiquitin or substrate protein

clashes with the RING0 domain, implying that this

domain must move in order for a substrate amino

group to access the active site.

Parkin activation

Recent studies have made tremendous progress in

understanding how parkin is activated. In vitro studies

of parkin have found that a wide variety of effectors

can promote parkin activity. These include point

mutations that disrupt inhibitory interdomain interac-

tions as well as N-terminal deletions [27–30]. The Ubl

domain plays a special role in parkin activation and a

number of binding partners that bind the Ubl domain

are associated with parkin activation [27]. The Ubl

domain recruits these binding partners such as the

SH3 domain and UIMs of endophilin A1, Eps15, pro-

teasomal subunits and ataxin-3 [31,35–37] through the

same hydrophobic surface centered around Ile44 that

it uses to interact with the RING1 domain, indicating

that the Ubl dissociates from RING1 upon activation.

PINK1 acts upstream of parkin and is required for

parkin activation and recruitment to depolarized mito-

chondria [12–15,47]. PINK1 phosphorylates parkin

Ubl domain on residue Ser65 to activate parkin [48–50]. Although the phospho-mimetic S65E mutation

stimulates parkin ligase activity in vitro, this mutation

is not able to bypass the PINK1 requirement for mito-

chondrial recruitment in cells and the non-phosphory-

latable S65A mutation is not completely impaired [48].

These observations led to the search for an additional

PINK1 substrate involved in the parkin activation and

the breakthrough discovery that ubiquitin can be phos-

phorylated by PINK1 [51–53]. Three groups concur-

rently observed that PINK1 can phosphorylate

ubiquitin on Ser65, the same position that is phos-

phorylated in the parkin Ubl domain. Further, they

showed that phosphorylated ubiquitin or phospho-

mimetic ubiquitin mutants (S65D/E) directly activate

parkin by enhancing the rate of E2–ubiquitin discharge

[51–53]. Parkin binds strongly to phospho-ubiquitin

with an affinity of 400 nM. Phosphorylation of Ser65

of parkin further increases the affinity 20-fold. In cells,

expression of the non-phosphorylatable S65A ubiqu-

itin delays parkin recruitment to the depolarized mito-

chondria, and mutation of both parkin and ubiquitin

at Ser65 abolishes parkin activation [53]. Further stud-

ies have revealed that PINK1 is not only a ubiquitin

kinase but is also capable of phosphorylating ubiquitin

in ubiquitin chains [25,54,55]. While phospho-ubiquitin

can still become activated by E1 and charged onto E2

enzymes, its activity in ubiquitination assays is E3

dependent [55]. Parkin shows somewhat less activity

with phospho-ubiquitin–E2 than with ubiquitin–E2[51]. Potentially more relevant is the observation that

phosphorylated ubiquitin chains were more resistant to

hydrolysis by 10 out of 12 deubiquitinases tested [55].

One possible explanation is the presence of a minor

conformation of phospho-ubiquitin that was detected

by NMR [55]. The minor conformation, corresponding

to about 30% of molecules, shows a b-strand slippage

that disrupts the Ile44 hydrophobic patch involved in

many ubiquitin interactions.

The structural details of parkin activation are

unknown but most of the evidence is compatible with

a simple two-state system. Inputs that activate parkin

shift the equilibrium between an inactive and active

conformation. There is no evidence of multiple steps

2081FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

M. Seirafi et al. Parkin structure and function

Page 7: Parkin structure and function

in the pathway: disruption of the RING0–RING2

interface increases ligase activity as does disruption of

the REP–RING1 interaction. The conformational

change between the two states has been suggested to

be a ‘butterfly’ movement in which parkin folds along

the axis between the RING0 and RING1 domains to

bring the two functional sites together [28]. Alterna-

tively, the active conformation could consist of an

ensemble of structures where the E2 binding and cata-

lytic sites transiently interact like beads on a string.

The release of the inhibitory interactions would suffice

to activate the RING1 and RING2 domains to carry

out the ubiquitin ligase activity.

The site of parkin phosphorylation is in close prox-

imity to the REP linker between IBR and RING2

domain in the autoinhibited conformation (Fig. 2B)

and could promote displacement of the REP linker

and Ubl domain to allow E2 binding. The site of

phospho-ubiquitin binding to parkin is also unknown

and it is not clear if the phospho-ubiquitin and phos-

phoUbl binding sites are the same or distinct. In one

of the crystal structures, a pocket formed by three

solvent exposed positively charged residues Lys161,

Arg163 and Lys211 in the RING0 domain was occu-

pied by a sulfate ion, suggesting a potential phos-

phate binding site [30]. While the PD-associated

mutants K161N and K211N fail to recruit to depo-

larized mitochondria [12–14], mutation of the basic

patch did not affect the binding affinity towards

phospho-ubiquitin, suggesting another role for this

pocket [25].

Parkin recruitment to mitochondria

In cultured cells, PINK1/parkin pathways can be acti-

vated by depolarizing mitochondria with uncouplers,

such as carbonyl cyanide m-chlorophenyl hydrazone

(CCCP). Following CCCP treatment, parkin shows a

very robust and complete recruitment to mitochondria

within several hours, which is then followed by the

clearance of mitochondria. Parkin recruitment to mito-

chondria requires PINK1 kinase activity; however,

there is no fixed stoichiometry between PINK1 levels

and parkin recruitment. This suggests that a molecule

other than PINK1 is recognized by parkin. This con-

trasts with the surprising observation that PINK1 arti-

ficially targeted to peroxisomes is sufficient to recruit

and activate parkin on peroxisomes [4]. A further

enigma is the requirement for parkin ubiquitin ligase

activity for its recruitment. Catalytically inactive

mutants of parkin do not show detectable recruitment

following mitochondrial depolarization [12,21].

The solution to this puzzle is now emerging with a

model of the events leading to the recruitment and

activation of parkin by phospho-ubiquitin (Fig. 4). In

the first step, the selective accumulation of PINK1 on

depolarized mitochondria leads to the phosphorylation

of low, basal levels of ubiquitin or parkin present on

mitochondria [25,51–53]. The system exhibits feedfor-

ward control as both ubiquitin and parkin phosphory-

lation are positive effectors of parkin ubiquitin ligase

activity [25,51–53]. Phosphorylation of ubiquitin

chains was recently shown to inhibit the action of

PINK1

Ub

UbUbUbUb

UbUb

UbUb

Phagophore

Mitophagy

PINK1 accumulation & activation

Phospho- ubiquitin

Parkin phosphorylation

Parkin recruitment and activation

Ubiquitination of mitochondrial proteins

Autophagy of mitochondria

Phosphorylation

PINK1

parkin

parkin

parkin

Depolarizedmitochondrion

Depolarizedmitochondrion

PINK1

A B

Fig. 4. Pathway of PINK1 activation of parkin leading to autophagy of depolarized mitochondria. (A) Flowchart of feedforward and feedback

activation of parkin. (B) Schematic of the quality control pathway. PINK1 accumulates on depolarized or damaged mitochondria and

phosphorylates ubiquitin and parkin present on the surface. Activated parkin produces additional ubiquitin chains on the mitochondria, which

in turn are phosphorylated by PINK1 to promote the recruitment of more parkin. The autophagosome forms around the heavily ubiquitinated

mitochondria, which are then eliminated by autophagy.

2082 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

Parkin structure and function M. Seirafi et al.

Page 8: Parkin structure and function

deubiquitinases [55], which would act as an additional

feedforward mechanism. Full activation of the system

requires a positive feedback loop where activation of

parkin increases the amount of mitochondrially conju-

gated ubiquitin, which is then phosphorylated by

PINK1 to recruit more parkin. The positive feedback

explains the requirement for both PINK1 and parkin

catalytic activities as well as how high levels of parkin

can be recruited and depleted from the cytosol by low,

endogenous levels of PINK1. In agreement with the

model, the requirement for parkin ligase activity can

be bypassed by using overexpressed tetra-ubiquitin

chains artificially targeted to mitochondria [56]. The

requirement for PINK1 activity can be further

bypassed by the use of phospho-mimetic chains for

S65E tetra-ubiquitin [54].

A number of questions remain unanswered. The

relative importance of the feedforward pathways is

unknown as is the order of the initial events. It is pos-

sible, although unlikely, that ubiquitin phosphoryla-

tion precedes chain formation and that parkin

incorporates phospho-ubiquitin directly into chains

[25,51,52]. The relative importance of mono-ubiquitin

and polyubiquitin chains is also unknown. Mono-

ubiquitin tethered to mitochondria is not sufficient to

recruit catalytically inactive parkin which suggests that

ubiquitin chains may play a special role [56].

Parkin/PINK1-mediated mitophagy inneurons

Although parkin recruitment to depolarized mitochon-

dria is a robust phenomenon in diverse mammalian

cell lines, it has been controversial whether mitophagy

is applicable in neurons and in PD pathogenesis. Con-

cerns are that most of these experiments use overex-

pressed parkin and that CCCP treatment leads to

rapid depolarization of the entire mitochondrial net-

work and non-physiological levels of damage to mito-

chondria, which probably is never the case in PD. In

experiments in primary neurons, parkin recruitment to

depolarized mitochondria is modest and only happens

after prolonged CCCP treatment or in the presence of

lysosomal or apoptosis inhibitors or special culture

conditions [57,58]. In part, this could arise from the

fact that most cell lines are glycolytic while neurons

are strictly dependent on oxidative phosphorylation

for mitochondrial ATP production; neuronal mito-

phagy studies may require more physiological methods

for induction of depolarized mitochondria. In a recent

paper, the short mitochondrial ARF protein was

shown to induce mitochondrial depolarization and

parkin/PINK1 autophagy both in cell lines and in

neurons [59]. Moreover, light activated ROS-induced

mitochondrial depolarization was also shown to initi-

ate parkin- and PINK1-dependent mitochondrial deg-

radation by autophagy in axonal mitochondria [60].

This confirms that parkin recruitment and activation

on mitochondria is relevant for PD.

Other functions of parkin

Parkin plays a number of roles outside of the induc-

tion of mitophagy. Parkin ubiquitination of outer

mitochondrial membrane proteins such as mitofusins,

optic atrophy 1 (OPA1) and Miro alters the balance of

fission to fusion and mitochondrial motility, facilitat-

ing the isolation of dysfunctional mitochondria from

the mitochondrial network for mitophagy [61]. Parkin

and PINK1 also work together to repair mildly dam-

aged mitochondria in response to mild oxidative stress

through the formation of mitochondria derived vesicles

(MDVs) enriched in oxidized proteins [62]. These

MDVs carry damaged cargo to lysosomes for degrada-

tion [63]. Complementary to mitophagy, parkin and

PINK1 balance the turnover of mitochondria by pro-

moting the synthesis of new mitochondria [64]. Over-

expressing parkin in proliferating cells and SH-SY5

cells increases mitochondrial transcription through

interaction with TFAM [65,66].

Parkin has been reported to promote cell survival

through various mechanisms although how parkin

becomes activated in these pathways is not always

clear. Parkin has been ascribed as preventing cell

death through proteosomal degradation of several pro-

teins such as parkin interacting substrate (PARIS),

aminoacyl-tRNA synthetase complex-interacting

multifunctional protein-2 (AIMP2) and Fbw7b, a sub-

strate-binding adaptor protein subunit of SCF E3

ubiquitin ligase complex [67–69]. Parkin may activate

prosurvival pathways by increasing nuclear factor jB(NF-jB) signaling [70] or decreasing activation of

c-jun N-terminal kinase [71–73]. A number of recent

papers describe interactions between parkin and Bcl

proteins in the mitochondrial apoptotic pathway

[69,74–77]. Parkin transcription is under the control of

p53, and reportedly mediates the Warburg effect of

p53 on glucose metabolism [78]. Parkin has also been

implicated in cell surface signaling by controlling epi-

dermal growth factor receptor internalization and Akt

signaling by ubiquitination of the endocytic scaffold

protein Eps15 [31]. Parkin is a tumor suppressor and

its inactivation has been reported in various human

cancers. Parkin is deleted in 30% of human tumors

and parkin-deficient mice are more susceptible to

tumorigenesis [78–82].

2083FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

M. Seirafi et al. Parkin structure and function

Page 9: Parkin structure and function

Parkin also has a key role in pathogen defense

through xenophagy, a pathway related to mitophagy.

In xenophagy, bacteria are marked with ubiquitin

chains that recruit ubiquitin-binding autophagy adap-

tors, leading to autophagosome formation and eventu-

ally fusion with the lysosome. The ubiquitinated

substrates and ligases involved in this pathway are

poorly understood. Genomic studies identified parkin

as a susceptibility factor for the intracellular bacterial

pathogen Mycobacterium leprae [83]. In a recent paper,

parkin has been shown to be required for resistance to

intracellular pathogens such as Mycobacterium tuber-

culosis and Salmonella enterica through an autophagy-

dependent mechanism [84]. The shared ancestry

between mitochondria and bacteria points to a com-

mon mechanism of parkin-mediated autophagy, but

whether PINK1 or a related kinase is required for xe-

nophagy is unknown.

Is parkin a good therapeutic target?

The recent progress in understanding the regulation of

parkin activity may seem appealing for new routes for

treating diseases with mitochondrial dysfunction. Par-

kin displays low basal activity and a small increase in

the activation of parkin could be sufficient to slow the

progression of PD in sporadic forms of the disease

where the wild-type protein is present [41]. Although

simplistic, small molecules that mimic phospho-ubiqu-

itin or disrupt autoinhibitory interactions might

enhance its neuroprotective action. In cultured cells,

mutation of Trp403 or Phe463 speeds recruitment of

parkin to mitochondria in a regulated process that

remains dependent on PINK1 and mitochondrial

depolarization [29]. A small molecule that binds tightly

to the pocket occupied by the amino acid side chains

would be expected to have the same effect. Alterna-

tively, the deubiquitinating (DUB) enzymes USP30

and USP15 were recently found to oppose parkin/

PINK1 in mitophagy, making inhibitors of these

DUBs prime candidates for drug design [85,86]. In

contrast, USP8 promotes parkin-mediated mitophagy

and thus agonists of this DUB could be developed

[26]. While it remains speculative with many chal-

lenges, the quality control pathway mediated by

PINK1 and parkin appears to offer multiple therapeu-

tic targets for the treatment of PD and other diseases

caused by dysfunctional mitochondria.

Acknowledgements

We thank Jean-Franc�ois Trempe and Xinlu Li for

reading the manuscript and constructive comments.

This work was supported by the Canadian Institutes

of Health Research grant MOP-125924 and student-

ships from the Fonds de la Recherche en Sant�e du

Qu�ebec in partnership with la Soci�et�e Parkinson du

Qu�ebec and the NSERC CREATE Training Program

in Bionanomachines.

Author contributions

MS, GK, and KG conceived and wrote the review.

References

1 Venderova K & Park DS (2012) Programmed cell death

in Parkinson’s disease. Cold Spring Harb Perspect Med

2, pii: a009365.

2 Valente EM, Abou-Sleiman PM, Caputo V, Muqit

MM, Harvey K, Gispert S, Ali Z, Del Turco D,

Bentivoglio AR, Healy DG et al. (2004) Hereditary

early-onset Parkinson’s disease caused by mutations in

PINK1. Science 304, 1158–1160.3 Narendra D, Walker JE & Youle R (2012)

Mitochondrial quality control mediated by PINK1 and

parkin: links to parkinsonism. Cold Spring Harb

Perspect Biol 4, pii: a011338.

4 Lazarou M, Jin SM, Kane LA & Youle RJ (2012)

Role of PINK1 binding to the TOM complex and

alternate intracellular membranes in recruitment

and activation of the E3 ligase parkin. Dev Cell 22,

320–333.5 Deas E, Plun-Favreau H, Gandhi S, Desmond H, Kjaer

S, Loh SHY, Renton AEM, Harvey RJ, Whitworth AJ,

Martins LM et al. (2011) PINK1 cleavage at position

A103 by the mitochondrial protease PARL. Hum Mol

Genet 20, 867–879.6 Greene AW, Grenier K, Aguileta MA, Muise S,

Farazifard R, Haque ME, McBride HM, Park DS &

Fon EA (2012) Mitochondrial processing peptidase

regulates PINK1 processing, import and parkin

recruitment. EMBO Rep 13, 378–385.7 Jin SM, Lazarou M, Wang CX, Kane LA,

Narendra DP & Youle RJ (2010) Mitochondrial

membrane potential regulates PINK1 import and

proteolytic destabilization by PARL. J Cell Biol 191,

933–942.8 Whitworth AJ, Lee JR, Ho VMW, Flick R,

Chowdhury R & McQuibban GA (2008) Rhomboid-7

and HtrA2/Omi act in a common pathway with the

Parkinson’s disease factors Pink1 and parkin. Dis

Model Mech 1, 168–174.9 Yamano K & Youle RJ (2013) PINK1 is degraded through

the N-end rule pathway. Autophagy 9, 1758–1769.10 Lin W & Kang UJ (2008) Characterization of PINK1

processing, stability, and subcellular localization.

J Neurochem 106, 464–474.

2084 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

Parkin structure and function M. Seirafi et al.

Page 10: Parkin structure and function

11 Zhou C, Huang Y, Shao YF, May J, Prou D, Perier

C, Dauer W, Schon EA & Przedborski S (2008) The

kinase domain of mitochondrial PINK1 faces the

cytoplasm. Proc Natl Acad Sci USA 105, 12022–12027.

12 Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier

CA, Shen J, Cookson MR & Youle RJ (2010) PINK1

is selectively stabilized on impaired mitochondria to

activate parkin. PLoS Biol 8, e1000298.

13 Geisler S, Holmstrom KM, Skujat D, Fiesel FC,

Rothfuss OC, Kahle PJ & Springer W (2010) PINK1/

Parkin-mediated mitophagy is dependent on VDAC1

and p62/SQSTM1. Nat Cell Biol 12, 119–131.14 Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K,

Gautier CA, Sou Y, Saiki S, Kawajiri S, Sato F et al.

(2010) PINK1 stabilized by mitochondrial

depolarization recruits parkin to damaged mitochondria

and activates latent parkin for mitophagy. J Cell Biol

189, 211–221.15 Vives-Bauza C, Zhou C, Huang Y, Cui M, de Vries

RLA, Kim J, May J, Tocilescu MA, Liu WC, Ko HS

et al. (2010) PINK1-dependent recruitment of parkin to

mitochondria in mitophagy. Proc Natl Acad Sci USA

107, 378–383.16 Hershko A & Ciechanover A (1998) The ubiquitin

system. Annu Rev Biochem 67, 425–479.17 Randow F & Youle RJ (2014) Self and nonself: how

autophagy targets mitochondria and bacteria. Cell Host

Microbe 15, 403–411.18 Komander D & Rape M (2012) The ubiquitin code.

Annu Rev Biochem 81, 203–229.19 Dye BT & Schulman BA (2007) Structural mechanisms

underlying posttranslational modification by ubiquitin-

like proteins. Annu Rev Biophys Biomol Struct 36, 131–150.

20 Berndsen CE & Wolberger C (2014) New insights into

ubiquitin E3 ligase mechanism. Nat Struct Mol Biol 21,

301–307.21 Lazarou M, Narendra DP, Jin SM, Tekle E, Banerjee S

& Youle RJ (2013) PINK1 drives parkin self-

association and HECT-like E3 activity upstream of

mitochondrial binding. J Cell Biol 200, 163–172.22 Wenzel DM, Lissounov A, Brzovic PS & Klevit RE

(2011) UBCH7 reactivity profile reveals parkin and

HHARI to be RING/HECT hybrids. Nature 474, 105–108.

23 Chan NC, Salazar AM, Pham AH, Sweredoski MJ,

Kolawa NJ, Graham RL, Hess S & Chan DC (2011)

Broad activation of the ubiquitin-proteasome system by

parkin is critical for mitophagy. Hum Mol Genet 20,

1726–1737.24 Sarraf SA, Raman M, Guarani-Pereira V, Sowa ME,

Huttlin EL, Gygi SP & Harper JW (2013) Landscape of

the PARKIN-dependent ubiquitylome in response to

mitochondrial depolarization. Nature 496, 372–376.

25 Ordureau A, Sarraf SA, Duda DM, Heo JM,

Jedrychowski MP, Sviderskiy VO, Olszewski JL,

Koerber JT, Xie T, Beausoleil SA et al. (2014)

Quantitative proteomics reveal a feedforward

mechanism for mitochondrial PARKIN translocation

and ubiquitin chain synthesis. Mol Cell 56, 360–375.26 Durcan TM, Tang MY, Perusse JR, Dashti EA,

Aguileta MA, McLelland GL, Gros P, Shaler TA,

Faubert D, Coulombe B et al. (2014) USP8 regulates

mitophagy by removing K6-linked ubiquitin conjugates

from parkin. EMBO J 33, 2473–2491.27 Chaugule VK, Burchell L, Barber KR, Sidhu A, Leslie

SJ, Shaw GS & Walden H (2011) Autoregulation of

parkin activity through its ubiquitin-like domain.

EMBO J 30, 2853–2867.28 Riley BE, Lougheed JC, Callaway K, Velasquez M,

Brecht E, Nguyen L, Shaler T, Walker D, Yang Y,

Regnstrom K et al. (2013) Structure and function of

parkin E3 ubiquitin ligase reveals aspects of RING and

HECT ligases. Nat Commun 4, 1982.

29 Trempe JF, Sauve V, Grenier K, Seirafi M, Tang MY,

Menade M, Al-Abdul-Wahid S, Krett J, Wong K,

Kozlov G et al. (2013) Structure of parkin reveals

mechanisms for ubiquitin ligase activation. Science 340,

1451–1455.30 Wauer T & Komander D (2013) Structure of the

human parkin ligase domain in an autoinhibited state.

EMBO J 32, 2099–2112.31 Fallon L, Belanger CML, Corera AT, Kontogiannea

M, Regan-Klapisz E, Moreau F, Voortman J, Haber

M, Rouleau G, Thorarinsdottir T et al. (2006) A

regulated interaction with the UIM protein Eps15

implicates parkin in EGF receptor trafficking and PI(3)

K-Akt signalling. Nat Cell Biol 8, 834–842.32 Spratt DE, Walden H & Shaw GS (2014) RBR E3

ubiquitin ligases: new structures, new insights, new

questions. Biochem J 458, 421–437.33 Cruts M, Theuns J & Van Broeckhoven C (2012)

Locus-specific mutation databases for

neurodegenerative brain diseases. Hum Mutat 33, 1340–1344.

34 Finney N, Walther F, Mantel PY, Stauffer D, Rovelli

G & Dev KK (2003) The cellular protein level of

parkin is regulated by its ubiquitin-like domain. J Biol

Chem 278, 16054–16058.35 Sakata E, Yamaguchi Y, Kurimoto E, Kikuchi J,

Yokoyama S, Yamada S, Kawahara H, Yokosawa H,

Hattori N, Mizuno Y et al. (2003) Parkin binds the

Rpn10 subunit of 26S proteasomes through its

ubiquitin-like domain. EMBO Rep 4, 301–306.36 Trempe JF, Chen CXQ, Grenier K, Camacho EM,

Kozlov G, McPherson PS, Gehring K & Fon EA

(2009) SH3 domains from a subset of BAR proteins

define a Ubl-binding domain and implicate parkin in

synaptic ubiquitination. Mol Cell 36, 1034–1047.

2085FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

M. Seirafi et al. Parkin structure and function

Page 11: Parkin structure and function

37 Bai JJ, Safadi SS, Mercier P, Barber KR & Shaw GS

(2013) Ataxin-3 is a multivalent ligand for the parkin

Ubl domain. Biochemistry 52, 7369–7376.38 Spratt DE, Martinez-Torres RJ, Noh YJ, Mercier P,

Manczyk N, Barber KR, Aguirre JD, Burchell L,

Purkiss A, Walden H et al. (2013) A molecular

explanation for the recessive nature of parkin-linked

Parkinson’s disease. Nat Commun 4, 1983.

39 Dove KK & Klevit RE (2013) Structural biology:

parkin’s serpentine shape revealed in the year of the

snake. Curr Biol 23, R691–R693.

40 Hristova VA, Beasley SA, Rylett RJ & Shaw GS (2009)

Identification of a novel Zn2+-binding domain in the

autosomal recessive juvenile parkinson-related E3 ligase

parkin. J Biol Chem 284, 14978–14986.41 Regnstrom K, Yan J, Nguyen L, Callaway K, Yang Y,

Diep L, Xing W, Adhikari A, Beroza P, Hom RK

et al. (2013) Label free fragment screening using surface

plasmon resonance as a tool for fragment finding -

analyzing parkin, a difficult CNS target. PLoS ONE 8,

e66879.

42 Beasley SA, Hristova VA & Shaw GS (2007) Structure

of the parkin in-between-ring domain provides insights

for E3-ligase dysfunction in autosomal recessive

Parkinson’s disease. Proc Natl Acad Sci USA 104,

3095–3100.43 Duda DM, Olszewski JL, Schuermann JP, Kurinov I,

Miller DJ, Nourse A, Alpi AF & Schulman BA (2013)

Structure of HHARI, a RING-IBR-RING ubiquitin

ligase: autoinhibition of an ariadne-family E3 and

insights into ligation mechanism. Structure 21, 1030–1041.

44 Stieglitz B, Rana RR, Koliopoulos MG, Morris-Davies

AC, Schaeffer V, Christodoulou E, Howell S, Brown

NR, Dikic I & Rittinger K (2013) Structural basis for

ligase-specific conjugation of linear ubiquitin chains by

HOIP. Nature 503, 422–426.45 Stieglitz B, Morris-Davies AC, Koliopoulos MG,

Christodoulou E & Rittinger K (2012) LUBAC

synthesizes linear ubiquitin chains via a thioester

intermediate. EMBO Rep 13, 840–846.46 Madegowda RH, Kishore A & Anand A (2005)

Mutational screening of the parkin gene among South

Indians with early onset Parkinson’s disease. J Neurol

Neurosurg Psychiatry 76, 1588–1590.47 Ziviani E, Tao RN & Whitworth AJ (2010) Drosophila

parkin requires PINK1 for mitochondrial translocation

and ubiquitinates Mitofusin. Proc Natl Acad Sci USA

107, 5018–5023.48 Iguchi M, Kujuro Y, Okatsu K, Koyano F, Kosako H,

Kimura M, Suzuki N, Uchiyama S, Tanaka K &

Matsuda N (2013) Parkin-catalyzed ubiquitin-ester

transfer is triggered by PINK1-dependent

phosphorylation. J Biol Chem 288, 22019–22032.

49 Okatsu K, Oka T, Iguchi M, Imamura K, Kosako H,

Tani N, Kimura M, Go E, Koyano F, Funayama M

et al. (2012) PINK1 autophosphorylation upon

membrane potential dissipation is essential for parkin

recruitment to damaged mitochondria. Nat Commun 3,

1016.

50 Kondapalli C, Kazlauskaite A, Zhang N, Woodroof

HI, Campbell DG, Gourlay R, Burchell L, Walden

H, Macartney TJ, Deak M et al. (2012) PINK1 is

activated by mitochondrial membrane potential

depolarization and stimulates parkin E3 ligase

activity by phosphorylating Serine 65. Open Biol 2,

120080.

51 Koyano F, Okatsu K, Kosako H, Tamura Y, Go E,

Kimura M, Kimura Y, Tsuchiya H, Yoshihara H,

Hirokawa T et al. (2014) Ubiquitin is phosphorylated

by PINK1 to activate parkin. Nature 510, 162–166.52 Kazlauskaite A, Kondapalli C, Gourlay R, Campbell

DG, Ritorto MS, Hofmann K, Alessi DR, Knebel A,

Trost M & Muqit MMK (2014) Parkin is activated by

PINK1-dependent phosphorylation of ubiquitin at Ser

(65). Biochem J 460, 127–139.53 Kane LA, Lazarou M, Fogel AI, Li Y, Yamano K,

Sarraf SA, Banerjee S & Youle RJ (2014) PINK1

phosphorylates ubiquitin to activate Parkin E3

ubiquitin ligase activity. J Cell Biol 205, 143–153.54 Shiba-Fukushima K, Arano T, Matsumoto G, Inoshita

T, Yoshida S, Ishihama Y, Ryu KY, Nukina N,

Hattori N & Imai Y (2014) Phosphorylation of

mitochondrial polyubiquitin by PINK1 promotes

parkin mitochondrial tethering. PLoS Genet 10,

e1004861.

55 Wauer T, Swatek KN, Wagstaff JL, Gladkova C,

Pruneda JN, Michel MA, Gersch M, Johnson CM,

Freund SM & Komander D (2015) Ubiquitin Ser65

phosphorylation affects ubiquitin structure, chain

assembly and hydrolysis. EMBO J, 34, 307–325.56 Zheng XD & Hunter T (2013) Parkin mitochondrial

translocation is achieved through a novel catalytic

activity coupled mechanism. Cell Res 23, 886–897.57 Grenier K, McLelland GL & Fon EA (2013) Parkin-

and PINK1-dependent mitophagy in neurons: will the

real pathway please stand up? Front Neurol 4, 100.

58 Cai Q, Zakaria HM, Simone A & Sheng ZH (2012)

Spatial parkin translocation and degradation of

damaged mitochondria via mitophagy in live cortical

neurons. Curr Biol 22, 545–552.59 Grenier K, Kontogiannea M & Fon EA (2014) Short

mitochondrial ARF triggers parkin/PINK1-dependent

mitophagy. J Biol Chem 289, 29519–29530.60 Ashrafi G, Schlehe JS, LaVoie MJ & Schwarz TL

(2014) Mitophagy of damaged mitochondria occurs

locally in distal neuronal axons and requires PINK1

and Parkin. J Cell Biol 206, 655–670.

2086 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

Parkin structure and function M. Seirafi et al.

Page 12: Parkin structure and function

61 Scarffe LA, Stevens DA, Dawson VL & Dawson TM

(2014) Parkin and PINK1: much more than mitophagy.

Trends Neurosci 37, 315–324.62 McLelland GL, Soubannier V, Chen CX, McBride HM

& Fon EA (2014) Parkin and PINK1 function in a

vesicular trafficking pathway regulating mitochondrial

quality control. EMBO J 33, 282–295.63 Soubannier V, McLelland GL, Zunino R, Braschi E,

Rippstein P, Fon EA & McBride HM (2012) A

vesicular transport pathway shuttles cargo from

mitochondria to lysosomes. Curr Biol 22, 135–141.64 Vincow ES, Merrihew G, Thomas RE, Shulman NJ,

Beyer RP, MacCoss MJ & Pallanck LJ (2013) The

PINK1-Parkin pathway promotes both mitophagy and

selective respiratory chain turnover in vivo. Proc Natl

Acad Sci USA 110, 6400–6405.65 Kuroda Y, Mitsui T, Kunishige M, Shono M, Akaike

M, Azuma H & Matsumoto T (2006) Parkin enhances

mitochondrial biogenesis in proliferating cells. Hum

Mol Genet 15, 883–895.66 Rothfuss O, Fischer H, Hasegawa T, Maisel M, Leitner

P, Miesel F, Sharma M, Bornemann A, Berg D, Gasser

T et al. (2009) Parkin protects mitochondrial genome

integrity and supports mitochondrial DNA repair. Hum

Mol Genet 18, 3832–3850.67 Shin JH, Ko HS, Kang H, Lee Y, Lee YI, Pletinkova

O, Troconso JC, Dawson VL & Dawson TM (2011)

PARIS (ZNF746) repression of PGC-1alpha

contributes to neurodegeneration in Parkinson’s disease.

Cell 144, 689–702.68 Lee Y, Karuppagounder SS, Shin JH, Lee YI, Ko HS,

Swing D, Jiang H, Kang SU, Lee BD, Kang HC et al.

(2013) Parthanatos mediates AIMP2-activated age-

dependent dopaminergic neuronal loss. Nat Neurosci

16, 1392–1400.69 Ekholm-Reed S, Goldberg MS, Schlossmacher MG &

Reed SI (2013) Parkin-dependent degradation of the F-

box protein Fbw7beta promotes neuronal survival in

response to oxidative stress by stabilizing Mcl-1. Mol

Cell Biol 33, 3627–3643.70 Muller-Rischart AK, Pilsl A, Beaudette P, Patra M,

Hadian K, Funke M, Peis R, Deinlein A, Schweimer C,

Kuhn PH et al. (2013) The E3 ligase parkin maintains

mitochondrial integrity by increasing linear

ubiquitination of NEMO. Mol Cell 49, 908–921.71 Hwang S, Kim D, Choi G, An SW, Hong YK, Suh

YS, Lee MJ & Cho KS (2010) Parkin suppresses c-Jun

N-Terminal kinase-induced cell death via transriptional

regulation in drosophila. Mol Cells 29, 575–580.72 Jiang HB, Ren Y, Zhao JH & Feng J (2004) Parkin

protects human dopaminergic neuroblastoma cells

against dopamine-induced apoptosis. Hum Mol Genet

13, 1745–1754.73 Ren Y, Jiang H, Yang F, Nakaso K & Feng J (2009)

Parkin protects dopaminergic neurons against

microtubule-depolymerizing toxins by attenuating

microtubule-associated protein kinase activation. J Biol

Chem 284, 4009–4017.74 Hollville E, Carroll RG, Cullen SP & Martin SJ (2014)

Bcl-2 family proteins participate in mitochondrial

quality control by regulating Parkin/PINK1-dependent

mitophagy. Mol Cell 55, 451–466.75 Johnson BN, Berger AK, Cortese GP & LaVoie MJ

(2012) The ubiquitin E3 ligase parkin regulates the

proapoptotic function of Bax. Proc Natl Acad Sci USA

109, 6283–6288.76 Berger AK, Cortese GP, Amodeo KD, Weihofen A,

Letai A & LaVoie MJ (2009) Parkin selectively alters

the intrinsic threshold for mitochondrial cytochrome c

release. Hum Mol Genet 18, 4317–4328.77 Zhang C, Lee S, Peng Y, Bunker E, Giaime E, Shen J,

Zhou Z & Liu X (2014) PINK1 triggers autocatalytic

activation of parkin to specify cell fate decisions. Curr

Biol 24, 1854–1865.78 Zhang C, Lin M, Wu R, Wang X, Yang B, Levine AJ,

Hu W & Feng Z (2011) Parkin, a p53 target gene,

mediates the role of p53 in glucose metabolism and the

Warburg effect. Proc Natl Acad Sci USA 108, 16259–16264.

79 Cesari R, Martin ES, Calin GA, Pentimalli F, Bichi R,

McAdams H, Trapasso F, Drusco A, Shimizu M,

Masciullo V et al. (2003) Parkin, a gene implicated in

autosomal recessive juvenile parkinsonism, is a

candidate tumor suppressor gene on chromosome 6q25-

q27. Proc Natl Acad Sci U S A 100, 5956–5961.80 Fujiwara M, Marusawa H, Wang HQ, Iwai A, Ikeuchi

K, Imai Y, Kataoka A, Nukina N, Takahashi R &

Chiba T (2008) Parkin as a tumor suppressor gene for

hepatocellular carcinoma. Oncogene 27, 6002–6011.81 Gong Y, Zack TI, Morris LG, Lin K, Hukkelhoven E,

Raheja R, Tan IL, Turcan S, Veeriah S, Meng S et al.

(2014) Pan-cancer genetic analysis identifies PARK2 as a

master regulator of G1/S cyclins. Nat Genet 46, 588–594.82 Veeriah S, Taylor BS, Meng S, Fang F, Yilmaz E,

Vivanco I, JanakiramanM, Schultz N, Hanrahan AJ, Pao

W et al. (2010) Somatic mutations of the Parkinson’s

disease-associated gene PARK2 in glioblastoma and other

human malignancies. Nat Genet 42, 77–82.83 Mira MT, Alcais A, Nguyen VT, Moraes MO, Di Flumeri

C, Vu HT, Mai CP, Nguyen TH, Nguyen NB, Pham XK

et al. (2004) Susceptibility to leprosy is associated with

PARK2 and PACRG.Nature 427, 636–640.84 Manzanillo PS, Ayres JS, Watson RO, Collins AC,

Souza G, Rae CS, Schneider DS, Nakamura K, Shiloh

MU & Cox JS (2013) The ubiquitin ligase parkin

mediates resistance to intracellular pathogens. Nature

501, 512–516.85 Bingol B, Tea JS, Phu L, Reichelt M, Bakalarski CE,

Song Q, Foreman O, Kirkpatrick DS & Sheng M

(2014) The mitochondrial deubiquitinase USP30

2087FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

M. Seirafi et al. Parkin structure and function

Page 13: Parkin structure and function

opposes parkin-mediated mitophagy. Nature 510, 370–375.

86 Cornelissen T, Haddad D, Wauters F, Van Humbeeck

C, Mandemakers W, Koentjoro B, Sue C, Gevaert K,

De Strooper B, Verstreken P et al. (2014) The

deubiquitinase USP15 antagonizes parkin-mediated

mitochondrial ubiquitination and mitophagy. Hum Mol

Genet 23, 5227–5242.

2088 FEBS Journal 282 (2015) 2076–2088 ª 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

Parkin structure and function M. Seirafi et al.